1
|
Pham TN, Schelling RE, Loh KH. Parkinson's disease and metabolic disorders, understanding their shared co-morbidity through the autonomic nervous system. ADVANCES IN GENETICS 2025; 113:199-247. [PMID: 40409798 DOI: 10.1016/bs.adgen.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and nonmotor dysfunctions. Its pathological hallmark is the aggregation of ɑ-synuclein in the central nervous system (CNS), leading to widespread loss of dopaminergic neurons in the substantia nigra (SN). Interestingly, metabolic disorders localized in the periphery, such as diabetes mellitus, frequently co-occur with PD. Emerging evidence highlights a bidirectional relationship: metabolic diseases may accelerate PD progression, while PD can exacerbate metabolic dysfunction. Beyond these associations, a growing body of research suggests that dysfunction in the peripheral nervous system, the primary communication bridge between the brain and peripheral organs, plays a critical role in these comorbidities. Autonomic nerve perturbation may accelerate dopaminergic neuronal loss in the SN and exacerbate metabolic dysregulation. This chapter synthesizes current evidence linking autonomic dysfunction with the progression of PD and related metabolic disorders, and it explores innovative therapeutic strategies leveraging this bidirectional relationship to address PD progression.
Collapse
Affiliation(s)
- Thanh N Pham
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Rebecca E Schelling
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Ken H Loh
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, United States; Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
2
|
Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024; 25:5277-5293. [PMID: 39558137 PMCID: PMC11624209 DOI: 10.1038/s44319-024-00321-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The central nervous system and adipose tissue interact through complex communication. This bidirectional signaling regulates metabolic functions. The hypothalamus, a key homeostatic brain region, integrates exteroceptive and interoceptive signals to control appetite, energy expenditure, glucose, and lipid metabolism. This regulation is partly achieved via the nervous modulation of white (WAT) and brown (BAT) adipose tissue. In this review, we highlight the roles of sympathetic and parasympathetic innervation in regulating WAT and BAT activities, such as lipolysis and thermogenesis. Adipose tissue, in turn, plays a dual role as an energy reservoir and an endocrine organ, secreting hormones that influence brain function and metabolic health. In addition, this review focuses on recently uncovered communication pathways, including extracellular vesicles and neuro-mesenchymal units, which add new layers of regulation and complexity to the brain-adipose tissue interaction. Finally, we also examine the consequences of disrupted communication between the brain and adipose tissue in metabolic disorders like obesity and type-2 diabetes, emphasizing the potential for new therapeutic strategies targeting these pathways to improve metabolic health.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile
- Physiology Department, Biological Science Faculty, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- IBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
3
|
Jarrah M, Tasabehji D, Fraer A, Mokadem M. Spinal afferent neurons: emerging regulators of energy balance and metabolism. Front Mol Neurosci 2024; 17:1479876. [PMID: 39582948 PMCID: PMC11583444 DOI: 10.3389/fnmol.2024.1479876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
Recent advancements in neurophysiology have challenged the long-held paradigm that vagal afferents serve as the primary conduits for physiological signals governing food intake and energy expenditure. An expanding body of evidence now illuminates the critical role of spinal afferent neurons in these processes, necessitating a reevaluation of our understanding of energy homeostasis regulation. This comprehensive review synthesizes cutting-edge research elucidating the multifaceted functions of spinal afferent neurons in maintaining metabolic equilibrium. Once predominantly associated with nociception and pathological states, these neurons are now recognized as integral components in the intricate network regulating feeding behavior, nutrient sensing, and energy balance. We explore the role of spinal afferents in food intake and how these neurons contribute to satiation signaling and meal termination through complex gut-brain axis pathways. The review also delves into the developing evidence that spinal afferents play a crucial role in energy expenditure regulation. We explore the ability of these neuronal fibers to carry signals that can modulate feeding behavior as well as adaptive thermogenesis in adipose tissue influencing basal metabolic rate, and thereby contributing to overall energy balance. This comprehensive analysis not only challenges existing paradigms but also opens new avenues for therapeutic interventions suggesting potential targets for treating metabolic disorders. In conclusion, this review highlights the need for a shift in our understanding of energy homeostasis, positioning spinal afferent neurons as key players in the intricate web of metabolic regulation.
Collapse
Affiliation(s)
- Mohammad Jarrah
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Dana Tasabehji
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Aviva Fraer
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Mohamad Mokadem
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Fraternal Orders of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
- Obesity Research and Education Initiative, University of Iowa, Iowa City, IA, United States
- Veterans Affairs Health Care System, Iowa City, IA, United States
| |
Collapse
|
4
|
Oliveira THCD, Gonçalves GKN. Effect of ovariectomy and high-fat diet on the expression of estrogen receptors and adipose tissue metabolism in wistar rats. Mol Cell Endocrinol 2024; 592:112327. [PMID: 38996834 DOI: 10.1016/j.mce.2024.112327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
This study addresses the increasing prevalence of obesity, especially among postmenopausal. Estrogen plays a crucial role in regulating adipose tissue in women, with its absence after menopause associated with metabolic complications. The study aimed to determine the lipolytic activity in different adipose tissue depots of ovariectomized rats submitted to a high-fat diet. Also, to analyze the expression of estrogen receptors in adipose tissues and perform histological and morphometric analyzes of these deposits. Female rats were ovariectomized (O) or sham operated (S). The animals were divided into groups: ovariectomized with high-fat diet (OF), sham-operated with high-fat diet (SF), ovariectomized with control diet (OC) or sham-operated with control diet as the control group (SC). After 24 weeks of consuming the diets, rats were killed and adipose tissue deposits were removed. Polymerase chain reaction was performed to analyze the expression of estrogen receptors in adipose tissues, lipolysis assay and histological analysis. Both the high-fat diet and ovariectomy increased body weight and adiposity. There was hypertrophy of adipocytes. Estrogen replacement therapy modulate lipolytic activity in different adipose depots, with different responses in relation to estrogen receptors. Estrogen receptor expression varied between fat depots. Mesenteric adipose tissue showed greater sensitivity to estrogen compared with others. Estrogen increased lipolytic activity in some fat depots, reducing in others. Expression of ERs depends of hormonal status and adipose tissue location, which may explain distinct actions of estrogen on the metabolism of adipose tissue and on the production of adipokines by them.
Collapse
|
5
|
Ryu V, Gumerova AA, Witztum R, Korkmaz F, Cullen L, Kannangara H, Moldavski O, Barak O, Lizneva D, Goosens KA, Stanley S, Kim SM, Yuen T, Zaidi M. An atlas of brain-bone sympathetic neural circuits in mice. eLife 2024; 13:e95727. [PMID: 38963696 PMCID: PMC11245306 DOI: 10.7554/elife.95727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
There is clear evidence that the sympathetic nervous system (SNS) mediates bone metabolism. Histological studies show abundant SNS innervation of the periosteum and bone marrow-these nerves consist of noradrenergic fibers that immunostain for tyrosine hydroxylase, dopamine beta-hydroxylase, or neuropeptide Y. Nonetheless, the brain sites that send efferent SNS outflow to the bone have not yet been characterized. Using pseudorabies (PRV) viral transneuronal tracing, we report, for the first time, the identification of central SNS outflow sites that innervate bone. We find that the central SNS outflow to bone originates from 87 brain nuclei, sub-nuclei, and regions of six brain divisions, namely the midbrain and pons, hypothalamus, hindbrain medulla, forebrain, cerebral cortex, and thalamus. We also find that certain sites, such as the raphe magnus (RMg) of the medulla and periaqueductal gray (PAG) of the midbrain, display greater degrees of PRV152 infection, suggesting that there is considerable site-specific variation in the levels of central SNS outflow to the bone. This comprehensive compendium illustrating the central coding and control of SNS efferent signals to bone should allow for a greater understanding of the neural regulation of bone metabolism, and importantly and of clinical relevance, mechanisms for central bone pain.
Collapse
Affiliation(s)
- Vitaly Ryu
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anisa Azatovna Gumerova
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ronit Witztum
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Hasni Kannangara
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ofer Moldavski
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Orly Barak
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ki A Goosens
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Sarah Stanley
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
6
|
Turner RT, Branscum AJ, Iwaniec UT. Long-duration leptin transgene expression in dorsal vagal complex does not alter bone parameters in female Sprague Dawley rats. Bone Rep 2024; 21:101769. [PMID: 38706522 PMCID: PMC11067478 DOI: 10.1016/j.bonr.2024.101769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
The hypothalamus and dorsal vagal complex (DVC) are both important for integration of signals that regulate energy balance. Increased leptin transgene expression in either the hypothalamus or DVC of female rats was shown to decrease white adipose tissue and circulating levels of leptin and adiponectin. However, in contrast to hypothalamus, leptin transgene expression in the DVC had no effect on food intake, circulating insulin, ghrelin and glucose, nor on thermogenic energy expenditure. These findings imply different roles for hypothalamus and DVC in leptin signaling. Leptin signaling is required for normal bone accrual and turnover. Leptin transgene expression in the hypothalamus normalized the skeletal phenotype of leptin-deficient ob/ob mice but had no long-duration (≥10 weeks) effects on the skeleton of leptin-replete rats. The goal of this investigation was to determine the long-duration effects of leptin transgene expression in the DVC on the skeleton of leptin-replete rats. To accomplish this goal, we analyzed bone from three-month-old female rats that were microinjected with recombinant adeno-associated virus encoding either rat leptin (rAAV-Leptin, n = 6) or green fluorescent protein (rAAV-GFP, control, n = 5) gene. Representative bones from the appendicular (femur) and axial (3rd lumbar vertebra) skeleton were evaluated following 10 weeks of treatment. Selectively increasing leptin transgene expression in the DVC had no effect on femur cortical or cancellous bone microarchitecture. Additionally, increasing leptin transgene expression had no effect on vertebral osteoblast-lined or osteoclast-lined bone perimeter or marrow adiposity. Taken together, the findings suggest that activation of leptin receptors in the DVC has minimal specific effects on the skeleton of leptin-replete female rats.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
7
|
Roy D, Dion E, Sepeda JA, Peng J, Lingam SR, Townsend K, Sas A, Sun W, Tedeschi A. α2δ1-mediated maladaptive sensory plasticity disrupts adipose tissue homeostasis following spinal cord injury. Cell Rep Med 2024; 5:101525. [PMID: 38663398 PMCID: PMC11148638 DOI: 10.1016/j.xcrm.2024.101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 01/13/2024] [Accepted: 04/02/2024] [Indexed: 05/23/2024]
Abstract
Spinal cord injury (SCI) increases the risk of cardiometabolic disorders, including hypertension, dyslipidemia, and insulin resistance. Not only does SCI lead to pathological expansion of adipose tissue, but it also leads to ectopic lipid accumulation in organs integral to glucose and insulin metabolism. The pathophysiological changes that underlie adipose tissue dysfunction after SCI are unknown. Here, we find that SCI exacerbates lipolysis in epididymal white adipose tissue (eWAT). Whereas expression of the α2δ1 subunit of voltage-gated calcium channels increases in calcitonin gene-related peptide-positive dorsal root ganglia neurons that project to eWAT, conditional deletion of the gene encoding α2δ1 in these neurons normalizes eWAT lipolysis after SCI. Furthermore, α2δ1 pharmacological blockade through systemic administration of gabapentin also normalizes eWAT lipolysis after SCI, preventing ectopic lipid accumulation in the liver. Thus, our study provides insight into molecular causes of maladaptive sensory processing in eWAT, facilitating the development of strategies to reduce metabolic and cardiovascular complications after SCI.
Collapse
Affiliation(s)
- Debasish Roy
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Elliot Dion
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jesse A Sepeda
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Juan Peng
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Sai Rishik Lingam
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kristy Townsend
- Department of Neurological Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Sas
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Wenjing Sun
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
8
|
Zhao JY, Zhou LJ, Ma KL, Hao R, Li M. MHO or MUO? White adipose tissue remodeling. Obes Rev 2024; 25:e13691. [PMID: 38186200 DOI: 10.1111/obr.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
In this review, we delve into the intricate relationship between white adipose tissue (WAT) remodeling and metabolic aspects in obesity, with a specific focus on individuals with metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO). WAT is a highly heterogeneous, plastic, and dynamically secreting endocrine and immune organ. WAT remodeling plays a crucial role in metabolic health, involving expansion mode, microenvironment, phenotype, and distribution. In individuals with MHO, WAT remodeling is beneficial, reducing ectopic fat deposition and insulin resistance (IR) through mechanisms like increased adipocyte hyperplasia, anti-inflammatory microenvironment, appropriate extracellular matrix (ECM) remodeling, appropriate vascularization, enhanced WAT browning, and subcutaneous adipose tissue (SWAT) deposition. Conversely, for those with MUO, WAT remodeling leads to ectopic fat deposition and IR, causing metabolic dysregulation. This process involves adipocyte hypertrophy, disrupted vascularization, heightened pro-inflammatory microenvironment, enhanced brown adipose tissue (BAT) whitening, and accumulation of visceral adipose tissue (VWAT) deposition. The review underscores the pivotal importance of intervening in WAT remodeling to hinder the transition from MHO to MUO. This insight is valuable for tailoring personalized and effective management strategies for patients with obesity in clinical practice.
Collapse
Affiliation(s)
- Jing Yi Zhao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Juan Zhou
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Le Ma
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Hao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Stavres J, Aultman RS, Brandner CF, Newsome TA, Vallecillo-Bustos A, Graybeal AJ. Fat-free mass is associated with exercise pressor responses, but not cold pressor responses, in humans: influence of maximal voluntary contraction. Front Sports Act Living 2024; 6:1352192. [PMID: 38510524 PMCID: PMC10952834 DOI: 10.3389/fspor.2024.1352192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Objective This study examined the contributions of fat mass (FM) and fat-free mass (FFM) to the magnitude of exercise pressor responses in humans. Methods The cumulative blood pressure responses (blood pressure index; BPI) to handgrip exercise (BPIhg), post-exercise-circulatory-occlusion (BPIpeco), and cold-pressor activation (BPIcpt) were collected from 67 individuals grouped by BMI (27.8 ± 7.3 kg/m2), FFM index (FFMi, 29.1 ± 3.8 kg/m2), and FM index (FMi 12.5 ± 4.8 kg/m2) quartiles. BPI responses to HG were also normalized to the time-tension index of HG, providing a relative index of exercise pressor response magnitude (BPInorm). Results BPIhg and BPIpeco were significantly elevated in the third FFMi quartile (p ≤ 0.034), while BPInorm significantly decreased in the second and fourth quartiles (p ≤ 0.029). In contrast, no differences in BPIcpt were observed across any FFMi, BMI, or FMi quartiles (p ≥ 0.268). FFM was independently associated with BPIhg, BPI-peco, and BPInorm (all p ≤ 0.049), however, FFM was eliminated as an independent predictor when maximal voluntary contraction (MVC) was included in these regression models (all p ≥ 0.495). Neither FFM nor MVC was associated with BPIcpt (p ≥ 0.229). Conclusions These findings indicate that exercise pressor responses, but not cold-pressor responses, are significantly associated with FFM in humans, and that this association is driven by FFM related differences in MVC.
Collapse
Affiliation(s)
- Jon Stavres
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Ryan S. Aultman
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Caleb F. Brandner
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
- Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Ta’Quoris A. Newsome
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Anabelle Vallecillo-Bustos
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Austin J. Graybeal
- School of Kinesiology and Nutrition, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
10
|
Wang Y, Ye L. The Afferent Function of Adipose Innervation. Diabetes 2024; 73:348-354. [PMID: 38377447 PMCID: PMC10882147 DOI: 10.2337/dbi23-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/18/2023] [Indexed: 02/22/2024]
Abstract
Adipose tissue innervation is critical for regulating metabolic and energy homeostasis. While the sympathetic efferent innervation of fat is well characterized, the role of sensory or afferent innervation remains less explored. This article reviews previous work on adipose innervation and recent advances in the study of sensory innervation of adipose tissues. We discuss key open questions, including the physiological implications of adipose afferents in homeostasis as well as potential cross talk with sympathetic neurons, the immune system, and hormonal pathways. We also outline the general technical challenges of studying dorsal root ganglia innervating fat, along with emerging technologies that may overcome these barriers. Finally, we highlight areas for further research to deepen our understanding of the afferent function of adipose innervation.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
11
|
Ryu V, Gumerova A, Witztum R, Korkmaz F, Kannangara H, Moldavski O, Barak O, Lizneva D, Goosens KA, Stanley S, Kim SM, Yuen T, Zaidi M. An Atlas of Brain-Bone Sympathetic Neural Circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579382. [PMID: 38370676 PMCID: PMC10871366 DOI: 10.1101/2024.02.07.579382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
There is clear evidence that the sympathetic nervous system (SNS) mediates bone metabolism. Histological studies show abundant SNS innervation of the periosteum and bone marrow--these nerves consist of noradrenergic fibers that immunostain for tyrosine hydroxylase, dopamine beta hydroxylase, or neuropeptide Y. Nonetheless, the brain sites that send efferent SNS outflow to bone have not yet been characterized. Using pseudorabies (PRV) viral transneuronal tracing, we report, for the first time, the identification of central SNS outflow sites that innervate bone. We find that the central SNS outflow to bone originates from 87 brain nuclei, sub-nuclei and regions of six brain divisions, namely the midbrain and pons, hypothalamus, hindbrain medulla, forebrain, cerebral cortex, and thalamus. We also find that certain sites, such as the raphe magnus (RMg) of the medulla and periaqueductal gray (PAG) of the midbrain, display greater degrees of PRV152 infection, suggesting that there is considerable site-specific variation in the levels of central SNS outflow to bone. This comprehensive compendium illustrating the central coding and control of SNS efferent signals to bone should allow for a greater understanding of the neural regulation of bone metabolism, and importantly and of clinical relevance, mechanisms for central bone pain.
Collapse
Affiliation(s)
- Vitaly Ryu
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ronit Witztum
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Hasni Kannangara
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ofer Moldavski
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Orly Barak
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ki A. Goosens
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sarah Stanley
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology (CeTMaP), Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
12
|
Azuara-Alvarez LE, Díaz-Muñoz M, Báez Ruiz A, Saderi N, Ramírez-Plascencia OD, Cárdenas-Romero S, Flores-Sandoval O, Salgado-Delgado R. Visceral fat sympathectomy ameliorates systemic and local stress response related to chronic sleep restriction. Exp Biol Med (Maywood) 2023; 248:2381-2392. [PMID: 38143435 PMCID: PMC10903249 DOI: 10.1177/15353702231214267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/23/2023] [Indexed: 12/26/2023] Open
Abstract
Disturbance of sleep homeostasis encompasses health issues, including metabolic disorders like obesity, diabetes, and augmented stress vulnerability. Sleep and stress interact bidirectionally to influence the central nervous system and metabolism. Murine models demonstrate that decreased sleep time is associated with an increased systemic stress response, characterized by endocrinal imbalance, including the elevated activity of hypothalamic-pituitary-adrenal axis, augmented insulin, and reduced adiponectin, affecting peripheral organs physiology, mainly the white adipose tissue (WAT). Within peripheral organs, a local stress response can also be activated by promoting the formation of corticosterone. This local amplifying glucocorticoid signaling is favored through the activation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). In WAT, 11β-HSD1 activity is upregulated by the sympathetic nervous system, suggesting a link between sleep loss, augmented stress response, and a potential WAT metabolic disturbance. To gain more understanding about this relationship, metabolic and stress responses of WAT-sympathectomized rats were analyzed to identify the contribution of the autonomic nervous system to stress response-related metabolic disorders during chronic sleep restriction. Male Wistar rats under sleep restriction were allowed just 6 h of daily sleep over eight weeks. Results showed that rats under sleep restriction presented higher serum corticosterone, increased adipose tissue 11β-HSD1 activity, weight loss, decreased visceral fat, augmented adiponectin, lower leptin levels, glucose tolerance impairment, and mildly decreased daily body temperature. In contrast, sympathectomized rats under sleep restriction exhibited decreased stress response (lower serum corticosterone and 11β-HSD1 activity). In addition, they maintained weight loss, explained by a reduced visceral fat pad, leptin, and adiponectin, improved glucose management, and persisting decline in body temperature. These results suggest autonomic nervous system is partially responsible for the WAT-exacerbated stress response and its metabolic and physiological disturbances.
Collapse
Affiliation(s)
- Lucia E Azuara-Alvarez
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Querétaro 76230, México
| | - Adrián Báez Ruiz
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Oscar Daniel Ramírez-Plascencia
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
- Neurology department, Beth Israel Deacones Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Skarleth Cárdenas-Romero
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
- Neurology department, Beth Israel Deacones Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Omar Flores-Sandoval
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Roberto Salgado-Delgado
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| |
Collapse
|
13
|
Lorsignol A, Rabiller L, Labit E, Casteilla L, Pénicaud L. The nervous system and adipose tissues: a tale of dialogues. Am J Physiol Endocrinol Metab 2023; 325:E480-E490. [PMID: 37729026 DOI: 10.1152/ajpendo.00115.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/16/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
White, beige, and brown adipose tissues play a crucial role in maintaining energy homeostasis. Due to the heterogeneous and diffuse nature of fat pads, this balance requires a fine and coordinated control of many actors and therefore permanent dialogues between these tissues and the central nervous system. For about two decades, many studies have been devoted to describe the neuro-anatomical and functional complexity involved to ensure this dialogue. Thus, if it is now clearly demonstrated that there is an efferent sympathetic innervation of different fat depots controlling plasticity as well as metabolic functions of the fat pad, the crucial role of sensory innervation capable of detecting local signals informing the central nervous system of the metabolic state of the relevant pads is much more recent. The purpose of this review is to provide the current state of knowledge on this subject.
Collapse
Affiliation(s)
- Anne Lorsignol
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Lise Rabiller
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Elodie Labit
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Louis Casteilla
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| | - Luc Pénicaud
- RESTORE, CNRS, Inserm, Université de Toulouse, Toulouse, France
| |
Collapse
|
14
|
Harris RBS. Low-dose peripheral leptin infusion produces selective activation of ventromedial hypothalamic and hindbrain STAT3. Am J Physiol Endocrinol Metab 2023; 325:E72-E82. [PMID: 37285599 PMCID: PMC10292972 DOI: 10.1152/ajpendo.00083.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 06/09/2023]
Abstract
Previous studies have shown that very low dose, acute, single peripheral leptin injections fully activate arcuate nucleus signal transducer and activator of transcription 3 (STAT3), but ventromedial hypothalamus (VMH) pSTAT3 continues to increase with higher doses of leptin that inhibit food intake. The lowest dose that inhibited intake increased circulating leptin 300-fold whereas food intake is inhibited by chronic peripheral leptin infusions that only double circulating leptin. This study examined whether the pattern of hypothalamic pSTAT3 was the same in leptin-infused rats as in leptin-injected rats. Male Sprague-Dawley rats received intraperitoneal infusions of 0, 5, 10, 20, or 40 µg leptin/day for 9 days. The highest dose of leptin increased serum leptin by 50-100%, inhibited food intake for 5 days, but inhibited weight gain and retroperitoneal fat mass for 9 days. Energy expenditure, respiratory exchange ratio, and brown fat temperature did not change. pSTAT3 was quantified in hypothalamic nuclei and the nucleus of the solitary tract (NTS) when food intake was inhibited and when it had returned to control levels. There was no effect of leptin on pSTAT3 in the medial or lateral arcuate nucleus or in the dorsomedial nucleus of the hypothalamus. VMH pSTAT3 was increased only at day 4 when food intake was inhibited, but NTS pSTAT3 was increased at both 4 and 9 days of infusion. These results suggest that activation of leptin VMH receptors contributes to the suppression of food intake, but that hindbrain receptors contribute to a sustained change in metabolism that maintains a reduced weight and fat mass.NEW & NOTEWORTHY Low-dose, chronic peripheral infusions of leptin produced an initial, transient inhibition of food intake that correlated with signal transducer and activator of transcription 3 (STAT3) activation in the ventromedial hypothalamus (VMH) and nucleus of the solitary tract (NTS). When intake normalized, but weight remained suppressed, the NTS was the only area that remained activated. These data suggest that leptin's primary function is to reduce body fat, that hypophagia is a means of achieving this and that different areas of the brain are responsible for the progressive response.
Collapse
Affiliation(s)
- Ruth B S Harris
- Center for Neuroinflammation and Cardiometabolic Disease, Georgia State University, Atlanta, Georgia, United States
| |
Collapse
|
15
|
Wang M, Geng X, Li K, Wang Y, Duan X, Hou C, Zhao L, Zhou H, Zhao D. Berberine ameliorates mesenteric vascular dysfunction by modulating perivascular adipose tissue in diet-induced obese in rats. BMC Complement Med Ther 2022; 22:198. [PMID: 35879716 PMCID: PMC9310483 DOI: 10.1186/s12906-022-03667-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Berberine (BBR) has been found to have antiobesity effects, and obesity can lead to adipose tissue degeneration. As a special adipose tissue, perivascular adipose tissue (PVAT) is closely related to vascular function and affects vasoconstriction and relaxation. What happens to PVAT in the early stages of diet-induced obesity and how BBR affects vascular function is the focus of our experimental study.
Methods
Sprague–Dawley rats were fed a high-fat diet (fat 34% kcal) for 4 weeks to simulate early obesity. Obese rats were treated with BBR (200 mg/kg) or metformin (MET, 100 mg/kg) by gavage for 2 weeks. The mesenteric arterioles were studied by atomic force microscopy (AFM). The force vs. time curves were observed and analysed to indicate vascular function. Nitric oxide (NO) and noradrenaline (NA) release was quantified using an organ bath with fluorescence assays and ELISA, respectively. Network pharmacology was used to analyse the overlapping targets related to BBR and obesity-related diseases, and the expression of NOS in mesenteric PVAT was further analysed with immunohistochemistry and real-time PCR. The serum inflammatory factor levels were tested.
Results
BBR significantly reduced the levels of blood glucose, blood lipids and inflammatory factors in serum. It also effectively improved abnormal mesenteric vasoconstriction and relaxation in obese rats. There was no significant change in mesenteric vascular structure, but NO production and eNOS expression were significantly increased in mesenteric PVAT (P < 0.01), and NA was decreased (P < 0.05) in obese rats. All these changes in the mesenteric arterioles and PVAT of obese rats were reversed by treatment with BBR and MET.
Conclusions
In diet-induced obesity in rats, the function of vasoconstriction and relaxation in mesenteric arterioles is altered, NO is increased, and NA is decreased in mesenteric PVAT. All these changes were reversed by BBR, suggesting a novel effect of BBR in ameliorating mesenteric vascular dysfunction by regulating PVAT.
Collapse
|
16
|
Hellmund C, Hepp P, Steinke H. The subpopliteal fat body. Ann Anat 2022; 245:151995. [PMID: 36182003 DOI: 10.1016/j.aanat.2022.151995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/25/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The knee is likely to get hurt due to its excessive weight-bearing, for which it is surrounded by strongly tensioned ligaments, connectives and muscles. These highly active structures are imbedded in fatty tissue. The Fatty and loose connective tissue of the knee recently gained a renaissance in research. While the Hoffa fat body in the ventral knee attracted attention over the last years, we have investigated a smaller, dorsal fat body, ventral to the popliteus muscle. This fat body has not been described before. MATERIALS AND METHODS 11 knees of 11 fresh specimens were investigated. All muscles but the popliteus muscle were removed. The popliteus was released from its tibial origin and dissected towards its tendinous insertion. Thereby, a subpopliteal fat body (SFB) was shown. The related vessels and nerves were evaluated. The size of the body was measured. Examples of histological slices were stained with HE and immunostained against neurofilament. RESULTS The SFB lies ventral of the popliteus muscle at the concave posterior tibia and attaches to the periosteum and the popliteus muscle. It is not attached to the posterior cruciate ligament. It is separated from the subpopliteal recess by a lamella deriving from the fibular head. Arterial and venous vessels are seen entering the SFB, deriving from the popliteal artery or the anterior tibial artery. A subbranch of the tibial nerve was seen to reach the SFB. The SFB could be identified in MRI scans and in plastinations. DISCUSSION Primarily, the SFB provides a gliding space for the mobile part of the popliteus muscle over the tibia. The SFB lies within the tibial concavity, ventral to the popliteus muscle. This is exactly where embryologically, the popliteal artery passes through, before its involution in later stages. Therefore, the SFB may show the former perivascular autonomic nerves which encompass the embryologically created arteries, from which we have seen the arterial remnants. The nerves seen here form neurovascular bundles which could be a source of pain, when compressed. This anatomy may explain the autonomic component of pain in the deep lateral region of the knee. The SFB is functional fat, comparable to the Hoffa's fat pad in the ventral knee.
Collapse
Affiliation(s)
- Christoph Hellmund
- Institut für Anatomie, Universität Leipzig, Liebigstr. 13, 04103 Leipzig.
| | - Pierre Hepp
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie und Plastische Chirurgie.
| | - Hanno Steinke
- Institut für Anatomie, Universität Leipzig, Liebigstr. 13, 04103 Leipzig.
| |
Collapse
|
17
|
Puente-Ruiz SC, Jais A. Reciprocal signaling between adipose tissue depots and the central nervous system. Front Cell Dev Biol 2022; 10:979251. [PMID: 36200038 PMCID: PMC9529070 DOI: 10.3389/fcell.2022.979251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
In humans, various dietary and social factors led to the development of increased brain sizes alongside large adipose tissue stores. Complex reciprocal signaling mechanisms allow for a fine-tuned interaction between the two organs to regulate energy homeostasis of the organism. As an endocrine organ, adipose tissue secretes various hormones, cytokines, and metabolites that signal energy availability to the central nervous system (CNS). Vice versa, the CNS is a critical regulator of adipose tissue function through neural networks that integrate information from the periphery and regulate sympathetic nerve outflow. This review discusses the various reciprocal signaling mechanisms in the CNS and adipose tissue to maintain organismal energy homeostasis. We are focusing on the integration of afferent signals from the periphery in neuronal populations of the mediobasal hypothalamus as well as the efferent signals from the CNS to adipose tissue and its implications for adipose tissue function. Furthermore, we are discussing central mechanisms that fine-tune the immune system in adipose tissue depots and contribute to organ homeostasis. Elucidating this complex signaling network that integrates peripheral signals to generate physiological outputs to maintain the optimal energy balance of the organism is crucial for understanding the pathophysiology of obesity and metabolic diseases such as type 2 diabetes.
Collapse
|
18
|
Kim MS. The neural basis of weight control and obesity. Exp Mol Med 2022; 54:347-348. [PMID: 35474337 PMCID: PMC9076921 DOI: 10.1038/s12276-022-00759-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Appetite Regulation Laboratory, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
19
|
González-Casacuberta I, Vilas D, Pont-Sunyer C, Tobías E, Cantó-Santos J, Valls-Roca L, García-García FJ, Garrabou G, Grau-Junyent JM, Martí MJ, Cardellach F, Morén C. Neuronal induction and bioenergetics characterization of human forearm adipose stem cells from Parkinson’s disease patients and healthy controls. PLoS One 2022; 17:e0265256. [PMID: 35290400 PMCID: PMC8923468 DOI: 10.1371/journal.pone.0265256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/26/2022] [Indexed: 11/18/2022] Open
Abstract
Neurodegenerative diseases, such as Parkinson’s disease, are heterogeneous disorders with a multifactorial nature involving impaired bioenergetics. Stem-regenerative medicine and bioenergetics have been proposed as promising therapeutic targets in the neurologic field. The rationale of the present study was to assess the potential of human-derived adipose stem cells (hASCs) to transdifferentiate into neuronal-like cells (NhASCs and neurospheres) and explore the hASC bioenergetic profile. hASC neuronal transdifferentiation was performed through neurobasal media and differentiation factor exposure. High resolution respirometry was assessed. Increased MAP-2 neuronal marker protein expression upon neuronal induction (p<0.05 undifferentiated hASCs vs. 28–36 days of differentiation) and increased bIII-tubulin neuronal marker protein expression upon neuronal induction (p<0.05 undifferentiated hASCs vs. 6-28-36 days of differentiation) were found. The bioenergetic profile was detectable through high-resolution respirometry approaches in hASCs but did not lead to differential oxidative capacity rates in healthy or clinically diagnosed PD-hASCs. We confirmed the capability of transdifferentiation to the neuronal-like profile of hASCs derived from the forearms of human subjects and characterized the bioenergetic profile. Suboptimal maximal respiratory capacity trends in PD were found. Neuronal induction leading to positive neuronal protein expression markers is a relevant issue that encourages the suitability of NhASC models in neurodegeneration.
Collapse
Affiliation(s)
- Ingrid González-Casacuberta
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Dolores Vilas
- Neurodegenerative Diseases Unit, Neurology Service, University Hospital Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Claustre Pont-Sunyer
- Neurology Unit, Hospital General de Granollers, Universitat Internacional de Catalunya, Barcelona, Catalonia, Spain
| | - Ester Tobías
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Judith Cantó-Santos
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Laura Valls-Roca
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Francesc Josep García-García
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Glòria Garrabou
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Josep Maria Grau-Junyent
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Maria Josep Martí
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Movement Disorders Unit, Neurology Service, Institut de Neurociències, University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Francesc Cardellach
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
| | - Constanza Morén
- Cellex-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine and Health Science, University of Barcelona, Spain
- Internal Medicine Department, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Madrid, Spain
- * E-mail:
| |
Collapse
|
20
|
Hilton C, Vasan SK, Neville MJ, Christodoulides C, Karpe F. The associations between body fat distribution and bone mineral density in the Oxford Biobank: a cross sectional study. Expert Rev Endocrinol Metab 2022; 17:75-81. [PMID: 34859739 PMCID: PMC8944227 DOI: 10.1080/17446651.2022.2008238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/16/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Body composition is associated with bone mineral density (BMD), but the precise associations between body fat distribution and BMD remain unclear. The regional adipose tissue depots have different metabolic profiles. We hypothesized that they would have independent associations with BMD. RESEARCH DESIGN AND METHODS We used data from 4,900 healthy individuals aged 30-50 years old from the Oxford Biobank to analyze associations between regional fat mass, lean mass and total BMD. RESULTS Total lean mass was strongly positively associated with BMD. An increase in total BMD was observed with increasing mass of all the fat depots, as measured either by anthropometry or DXA, when accounting for lean mass. However, on adjustment for both total fat mass and lean mass, fat depot specific associations emerged. Increased android and visceral adipose tissue mass in men, and increased visceral adipose tissue mass in women, were associated with lower BMD. CONCLUSIONS Fat distribution alters the association between adiposity and BMD.
Collapse
Affiliation(s)
- Catriona Hilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Senthil K Vasan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Trust, Oxford, UK
| | - Constantinos Christodoulides
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Trust, Oxford, UK
| |
Collapse
|
21
|
Dalmasso C, Leachman JR, Ghuneim S, Ahmed N, Schneider ER, Thibault O, Osborn JL, Loria AS. Epididymal Fat-Derived Sympathoexcitatory Signals Exacerbate Neurogenic Hypertension in Obese Male Mice Exposed to Early Life Stress. Hypertension 2021; 78:1434-1449. [PMID: 34601958 PMCID: PMC8516729 DOI: 10.1161/hypertensionaha.121.17298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/19/2021] [Indexed: 01/06/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| | - Jacqueline R. Leachman
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| | - Sundus Ghuneim
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| | - Nermin Ahmed
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| | - Eve R. Schneider
- Department of Biology, College of Arts and Sciences (E.R.S., J.L.O.), University of Kentucky, Lexington
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| | - Jeffrey L. Osborn
- Department of Biology, College of Arts and Sciences (E.R.S., J.L.O.), University of Kentucky, Lexington
| | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, College of Medicine (C.D., J.R.L., S.G., N.A., O.T., A.S.L.), University of Kentucky, Lexington
| |
Collapse
|
22
|
O'Brien CJO, Haberman ER, Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol 2021; 37:549-573. [PMID: 34613819 PMCID: PMC7614880 DOI: 10.1146/annurev-cellbio-120319-114106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Emma R Haberman
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| |
Collapse
|
23
|
Overexpression of the Gene Encoding Neurosecretory Protein GL Precursor Prevents Excessive Fat Accumulation in the Adipose Tissue of Mice Fed a Long-Term High-Fat Diet. Molecules 2021; 26:molecules26196006. [PMID: 34641550 PMCID: PMC8512635 DOI: 10.3390/molecules26196006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
We previously identified a novel small hypothalamic protein, neurosecretory protein GL (NPGL), which induces feeding behavior and fat accumulation in rodents depending on their diet. In the present study, we explored the effects of NPGL on feeding behavior and energy metabolism in mice placed on a long-term high-fat diet with 60% calories from fat (HFD 60). Overexpression of the NPGL precursor gene (Npgl) over 18 weeks increased food intake and weight. The weekly weight gain of Npgl-overexpressing mice was higher than that of controls until 7 weeks from induction of overexpression, after which it ceased to be so. Oral glucose tolerance tests showed that Npgl overexpression maintained glucose tolerance and increased blood insulin levels, and intraperitoneal insulin tolerance tests showed that it maintained insulin sensitivity. At the experimental endpoint, Npgl overexpression was associated with increased mass of the perirenal white adipose tissue (WAT) and decreased mass of the epididymal WAT (eWAT), resulting in little effect on the total WAT mass. These results suggest that under long-term HFD 60 feeding, Npgl overexpression may play a role in avoiding metabolic disturbance both by accelerating energy storage and by suppressing excess fat accumulation in certain tissues, such as the eWAT.
Collapse
|
24
|
Zhang Y, Zhou L, Lian H, Zhang Y, Tong S, Wang Z. Dopamine receptor 2 downregulation and brain-derived neurotrophic factor upregulation in the paraventricular nucleus are correlated with brown adipose tissue thermogenesis in rats with bilateral substantia nigra lesions. J Chem Neuroanat 2021; 117:102016. [PMID: 34454019 DOI: 10.1016/j.jchemneu.2021.102016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/19/2023]
Abstract
The thermogenesis resulting from brown adipose tissue (BAT)-induced energy consumption is an important method of energy regulation. It has been reported that brain-derived neurotrophic factor (BDNF)-positive neurons in the paraventricular nucleus (PVN) can regulate adaptive thermogenesis in interscapular brown adipose tissue (IBAT), but the upstream regulatory mechanism is still unclear. Our previous studies have found that a large number of dopamine (DA) receptors (DRs) are expressed on BDNF-positive neurons in the PVN and that the substantia nigra (SN) can directly project to the PVN (forming the SN-PVN pathway). Therefore, we speculate that DA in the SN can regulate the expression of BDNF via DRs and then affect IBAT thermogenesis. In this study, bilateral SN lesions were induced in rats with 6-hydroxydopamine (6-OHDA), and the altered expression of DRs and BDNF in the PVN and the metabolic changes in IBAT were studied via double immunofluorescence and western blotting. The results showed that BDNF-positive neurons in the PVN expressed DR 1 (D1) and DR 2 (D2) and were surrounded by a large number of tyrosine hydroxylase (TH)-positive nerve fibers. Compared with the control group, the 6-OHDA group exhibited significantly fewer TH-positive neurons and significantly lower TH expression in the SN, but body weight, IBAT weight and food consumption did not differ between the groups. In the PVN, BDNF expression was upregulated in the 6-OHDA group, while D2 and TH expression was downregulated. In IBAT, the expression of uncoupling protein-1 (UCP-1), phosphorylated hormone-sensitive lipase (p-HSL), TH and β3-adrenergic receptor (β3-AR) was increased, while the expression of fatty acid synthase (FAS) was decreased. The IBAT cell diameter was also decreased in the 6-OHDA group. The results suggest that the SN-PVN pathway may be an upstream neural pathway that can affect BDNF expression in the PVN and that DRs may mediate its regulatory effects. This study expands our understanding of the relationship between DA and obesity.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Li Zhou
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Hui Lian
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yimin Zhang
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Shilin Tong
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhiyong Wang
- Department of Human Anatomy and Histoembrology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang 453003, China; Henan International Joint Laboratory of Noninvasive Neuromodulation, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
25
|
Lee HJ, Jin BY, Park MR, Seo KS, Jeong YT, Choi SH, Kim DH. Deletion of adipose triglyceride lipase abolishes blood flow increase after β3-adrenergic stimulation in visceral adipose tissue of mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2021; 25:355-363. [PMID: 34187952 PMCID: PMC8255121 DOI: 10.4196/kjpp.2021.25.4.355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Hye-Jin Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Bo-Yeong Jin
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Mi-Rae Park
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Kwan Sik Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Yong Taek Jeong
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
26
|
Gomes A, Leite F, Ribeiro L. Adipocytes and macrophages secretomes coregulate catecholamine-synthesizing enzymes. Int J Med Sci 2021; 18:582-592. [PMID: 33437193 PMCID: PMC7797554 DOI: 10.7150/ijms.52219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity associates with macrophage accumulation in adipose tissue where these infiltrating cells interact with adipocytes and contribute to the systemic chronic metabolic inflammation present in immunometabolic diseases. Tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT) are two of the main enzymes of catecholamines (CA) synthesis. Adipocytes and macrophages produce, secrete and respond to CA, but the regulation of their synthesis in the interplay between immune and metabolic systems remains unknown. A model of indirect cell coculture with conditioned medium (CM) from RAW 264.7 macrophages with or without LPS-activation and 3T3-L1 adipocytes and preadipocytes was established to study the effect of cellular secretomes on the expression of the above enzymes. During the adipocyte differentiation process, we found a decrease of TH and PNMT expression. The secretome from LPS-activated macrophages downregulated TH and PNMT expression in preadipocytes, but not in mature adipocytes. Mature adipocytes CM induced a decrease of PNMT levels in RAW 264.7 macrophages. Pre and mature adipocytes showed a similar pattern of TH, PNMT and peroxisome proliferator-activated receptor gamma expression after exposure to pro and anti-inflammatory cytokines. We evidenced macrophages and adipocytes coregulate the expression of CA synthesis enzymes through secretome, with non-inflammatory signaling networks possibly being involved. Mediators released by macrophages seem to equally affect CA production by adipocytes, while adipocytes secretome preferentially affect AD production by macrophages. CA synthesis seems to be more determinant in early stages of adipogenic differentiation. Our results suggest that CA are key signaling molecules in the regulation of immune-metabolic crosstalk within the adipose tissue.
Collapse
Affiliation(s)
- Andreia Gomes
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto. Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto. Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Fernanda Leite
- Department of Clinical Haematology, Centro Hospitalar Universitário of Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
- UMIB/ICBAS - Unit for Multidisciplinary Investigation in Biomedicine- Institutode Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Laura Ribeiro
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto. Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto. Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto. Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
27
|
Blaszkiewicz M, Wood E, Koizar S, Willows J, Anderson R, Tseng YH, Godwin J, Townsend KL. The involvement of neuroimmune cells in adipose innervation. Mol Med 2020; 26:126. [PMID: 33297933 PMCID: PMC7727151 DOI: 10.1186/s10020-020-00254-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Innervation of adipose tissue is essential for the proper function of this critical metabolic organ. Numerous surgical and chemical denervation studies have demonstrated how maintenance of brain-adipose communication through both sympathetic efferent and sensory afferent nerves helps regulate adipocyte size, cell number, lipolysis, and 'browning' of white adipose tissue. Neurotrophic factors are growth factors that promote neuron survival, regeneration, and plasticity, including neurite outgrowth and synapse formation. Peripheral immune cells have been shown to be a source of neurotrophic factors in humans and mice. Although a number of immune cells reside in the adipose stromal vascular fraction (SVF), it has remained unclear what roles they play in adipose innervation. We previously demonstrated that adipose SVF secretes brain derived neurotrophic factor (BDNF). METHODS We now show that deletion of this neurotrophic factor from the myeloid lineage of immune cells led to a 'genetic denervation' of inguinal subcutaneous white adipose tissue (scWAT), thereby causing decreased energy expenditure, increased adipose mass, and a blunted UCP1 response to cold stimulation. RESULTS We and others have previously shown that noradrenergic stimulation via cold exposure increases adipose innervation in the inguinal depot. Here we have identified a subset of myeloid cells that home to scWAT upon cold exposure and are Ly6C+ CCR2+ Cx3CR1+ monocytes/macrophages that express noradrenergic receptors and BDNF. This subset of myeloid lineage cells also clearly interacted with peripheral nerves in the scWAT and were therefore considered neuroimmune cells. CONCLUSIONS We propose that these myeloid lineage, cold induced neuroimmune cells (CINCs) are key players in maintaining adipose innervation as well as promoting adipose nerve remodeling under noradrenergic stimulation, such as cold exposure.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- School of Biology and Ecology, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Elizabeth Wood
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Sigi Koizar
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Jake Willows
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Ryan Anderson
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - James Godwin
- Jackson Laboratory, Bar Harbor, ME, USA
- MDI Biological Laboratory, Bar Harbor, ME, USA
| | - Kristy L Townsend
- School of Biology and Ecology, University of Maine, Orono, ME, USA.
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA.
- The Ohio State University, 1014 Biomedical Research Tower, 460 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Bastías-Pérez M, Serra D, Herrero L. Dietary Options for Rodents in the Study of Obesity. Nutrients 2020; 12:nu12113234. [PMID: 33105762 PMCID: PMC7690621 DOI: 10.3390/nu12113234] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/05/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity and its associated metabolic diseases are currently a priority research area. The increase in global prevalence at different ages is having an enormous economic and health impact. Genetic and environmental factors play a crucial role in the development of obesity, and diet is one of the main factors that contributes directly to the obesogenic phenotype. Scientific evidence has shown that increased fat intake is associated with the increase in body weight that triggers obesity. Rodent animal models have been extremely useful in the study of obesity since weight gain can easily be induced with a high-fat diet. Here, we review the dietary patterns and physiological mechanisms involved in the dynamics of energy balance. We report the main dietary options for the study of obesity and the variables to consider in the use of a high-fat diet, and assess the progression of obesity and diet-induced thermogenesis.
Collapse
Affiliation(s)
- Marianela Bastías-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; (M.B.-P.); (D.S.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
29
|
Martínez-Sánchez N. There and Back Again: Leptin Actions in White Adipose Tissue. Int J Mol Sci 2020; 21:ijms21176039. [PMID: 32839413 PMCID: PMC7503240 DOI: 10.3390/ijms21176039] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Leptin is a hormone discovered almost 30 years ago with important implications in metabolism. It is primarily produced by white adipose tissue (WAT) in proportion to the amount of fat. The discovery of leptin was a turning point for two principle reasons: on one hand, it generated promising expectations for the treatment of the obesity, and on the other, it changed the classical concept that white adipose tissue was simply an inert storage organ. Thus, adipocytes in WAT produce the majority of leptin and, although its primary role is the regulation of fat stores by controlling lipolysis and lipogenesis, this hormone also has implications in other physiological processes within WAT, such as apoptosis, browning and inflammation. Although a massive number of questions related to leptin actions have been answered, the necessity for further clarification facilitates constantly renewing interest in this hormone and its pathways. In this review, leptin actions in white adipose tissue will be summarized in the context of obesity.
Collapse
|
30
|
Pralle RS, Schultz NE, White HM, Weigel KA. Hyperketonemia GWAS and parity-dependent SNP associations in Holstein dairy cows intensively sampled for blood β-hydroxybutyrate concentration. Physiol Genomics 2020; 52:347-357. [PMID: 32628084 DOI: 10.1152/physiolgenomics.00016.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hyperketonemia (HYK) is a metabolic disorder that affects early postpartum dairy cows; however, there has been limited success in identifying genomic variants contributing to HYK susceptibility. We conducted a genome-wide association study (GWAS) using HYK phenotypes based on an intensive screening protocol, interrogated genotype interactions with parity group (GWIS), and evaluated the enrichment of annotated metabolic pathways. Holstein cows were enrolled into the experiment after parturition, and blood samples were collected at four timepoints between 5 and 18 days postpartum. Concentration of blood β-hydroxybutyrate (BHB) was quantified cow-side via a handheld BHB meter. Cows were labeled as a HYK case when at least one blood sample had BHB ≥ 1.2 mmol/L, and all other cows were considered non-HYK controls. After quality control procedures, 1,710 cows and 58,699 genotypes were available for further analysis. The GWAS and GWIS were performed using the forward feature select linear mixed model method. There was evidence for an association between ARS-BFGL-NGS-91238 and HYK susceptibility, as well as parity-dependent associations to HYK for BovineHD0600024247 and BovineHD1400023753. Candidate genes annotated to these single nuclear polymorphism associations have been previously associated with obesity, diabetes, insulin resistance, and fatty liver in humans and rodent models. Enrichment analysis revealed focal adhesion and axon guidance as metabolic pathways contributing to HYK etiology, while genetic variation in pathways related to insulin secretion and sensitivity may affect HYK susceptibility in a parity-dependent matter. In conclusion, the present work proposes several novel marker associations and metabolic pathways contributing to genetic risk for HYK susceptibility.
Collapse
Affiliation(s)
- Ryan S Pralle
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nichol E Schultz
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heather M White
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kent A Weigel
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
31
|
Review: Following the smoke signals: inflammatory signaling in metabolic homeostasis and homeorhesis in dairy cattle. Animal 2020; 14:s144-s154. [PMID: 32024563 DOI: 10.1017/s1751731119003203] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammatory cascades are a critical component of the immune response to infection or tissue damage, involving an array of signals, including water-soluble metabolites, lipid mediators and several classes of proteins. Early investigation of these signaling pathways focused largely on immune cells and acute disease models. However, more recent findings have highlighted critical roles of both immune cells and inflammatory mediators on tissue remodeling and metabolic homeostasis in healthy animals. In dairy cattle, inflammatory signals in various tissues and in circulation change rapidly and dramatically, starting just prior to and at the onset of lactation. Furthermore, several observations in healthy cows point to homeostatic control of inflammatory tone, which we define as a regulatory process to balance immune tolerance with activation to keep downstream effects under control. Recent evidence suggests that peripartum inflammatory changes influence whole-body nutrient flux of dairy cows over the course of days and months. Inflammatory mediators can suppress appetite, even at levels that do not induce acute responses (e.g. fever), thereby decreasing nutrient availability. On the other hand, inhibition of inflammatory signaling with non-steroidal anti-inflammatory drug (NSAID) treatment suppresses hepatic gluconeogenesis, leading to hypoglycemia in some cases. Over the long term, though, peripartum NSAID treatment substantially increases peak and whole-lactation milk synthesis by multiparous cows. Inflammatory regulation of nutrient flux may provide a homeorhetic mechanism to aid cows in adapting to rapid changes in metabolic demand at the onset of lactation, but excessive systemic inflammation has negative effects on metabolic homeostasis through inhibition of appetite and promotion of immune cell activity. Thus, in this review, we provide perspectives on the overlapping regulation of immune responses and metabolism by inflammatory mediators, which may provide a mechanistic underpinning for links between infectious and metabolic diseases in transition dairy cows. Moreover, we point to novel approaches to the management of this challenging phase of the production cycle.
Collapse
|
32
|
Metabolic regulation and the anti-obesity perspectives of brown adipose tissue (BAT); a systematic review. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.obmed.2019.100163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Abstract
Neuroimmunology and immunometabolism are burgeoning topics of study, but the intersection of these two fields is scarcely considered. This interplay is particularly prevalent within adipose tissue, where immune cells and the sympathetic nervous system (SNS) have an important role in metabolic homeostasis and pathology, namely in obesity. In the present Review, we first outline the established reciprocal adipose-SNS relationship comprising the neuroendocrine loop facilitated primarily by adipose tissue-derived leptin and SNS-derived noradrenaline. Next, we review the extensive crosstalk between adipocytes and resident innate immune cells as well as the changes that occur in these secretory and signalling pathways in obesity. Finally, we discuss the effect of SNS adrenergic signalling in immune cells and conclude with exciting new research demonstrating an immutable role for SNS-resident macrophages in modulating SNS-adipose crosstalk. We posit that the latter point constitutes the existence of a new field - neuroimmunometabolism.
Collapse
Affiliation(s)
- Chelsea M Larabee
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Oliver C Neely
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Ana I Domingos
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK.
- The Howard Hughes Medical Institute (HHMI), New York, NY, USA.
| |
Collapse
|
34
|
Dalmasso C, Leachman JR, Osborn JL, Loria AS. Sensory signals mediating high blood pressure via sympathetic activation: role of adipose afferent reflex. Am J Physiol Regul Integr Comp Physiol 2019; 318:R379-R389. [PMID: 31868518 DOI: 10.1152/ajpregu.00079.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Blood pressure regulation in health and disease involves a balance between afferent and efferent signals from multiple organs and tissues. Although there are numerous reviews focused on the role of sympathetic nerves in different models of hypertension, few have revised the contribution of afferent nerves innervating adipose tissue and their role in the development of obesity-induced hypertension. Both clinical and basic research support the beneficial effects of bilateral renal denervation in lowering blood pressure. However, recent studies revealed that afferent signals from adipose tissue, in an adipose-brain-peripheral pathway, could contribute to the increased sympathetic activation and blood pressure during obesity. This review focuses on the role of adipose tissue afferent reflexes and briefly describes a number of other afferent reflexes modulating blood pressure. A comprehensive understanding of how multiple afferent reflexes contribute to the pathophysiology of essential and/or obesity-induced hypertension may provide significant insights into improving antihypertensive therapeutic approaches.
Collapse
Affiliation(s)
- Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jacqueline R Leachman
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jeffrey L Osborn
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
35
|
Blaszkiewicz M, Willows JW, Dubois AL, Waible S, DiBello K, Lyons LL, Johnson CP, Paradie E, Banks N, Motyl K, Michael M, Harrison B, Townsend KL. Neuropathy and neural plasticity in the subcutaneous white adipose depot. PLoS One 2019; 14:e0221766. [PMID: 31509546 PMCID: PMC6738614 DOI: 10.1371/journal.pone.0221766] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 08/14/2019] [Indexed: 12/30/2022] Open
Abstract
The difficulty in obtaining as well as maintaining weight loss, together with the impairment of metabolic control in conditions like diabetes and cardiovascular disease, may represent pathological situations of inadequate neural communication between the brain and peripheral organs and tissues. Innervation of adipose tissues by peripheral nerves provides a means of communication between the master metabolic regulator in the brain (chiefly the hypothalamus), and energy-expending and energy-storing cells in the body (primarily adipocytes). Although chemical and surgical denervation studies have clearly demonstrated how crucial adipose tissue neural innervation is for maintaining proper metabolic health, we have uncovered that adipose tissue becomes neuropathic (ie: reduction in neurites) in various conditions of metabolic dysregulation. Here, utilizing both human and mouse adipose tissues, we present evidence of adipose tissue neuropathy, or loss of proper innervation, under pathophysiological conditions such as obesity, diabetes, and aging, all of which are concomitant with insult to the adipose organ as well as metabolic dysfunction. Neuropathy is indicated by loss of nerve fiber protein expression, reduction in synaptic markers, and lower neurotrophic factor expression in adipose tissue. Aging-related adipose neuropathy particularly results in loss of innervation around the tissue vasculature, which cannot be reversed by exercise. Together with indications of neuropathy in muscle and bone, these findings underscore that peripheral neuropathy is not restricted to classic tissues like the skin of distal extremities, and that loss of innervation to adipose may trigger or exacerbate metabolic diseases. In addition, we have demonstrated stimulation of adipose tissue neural plasticity with cold exposure, which may ameliorate adipose neuropathy and be a potential therapeutic option to re-innervate adipose and restore metabolic health.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono ME, United States of America
| | - Jake W. Willows
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
| | - Amanda L. Dubois
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono ME, United States of America
| | - Stephen Waible
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
| | - Kristen DiBello
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
| | - Lila L. Lyons
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
| | - Cory P. Johnson
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono ME, United States of America
| | - Emma Paradie
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
| | - Nicholas Banks
- Maine Medical Center Research Institute, Scarborough ME, United States of America
| | - Katherine Motyl
- Maine Medical Center Research Institute, Scarborough ME, United States of America
| | - Merilla Michael
- University of New England, Biddeford ME, United States of America
| | | | - Kristy L. Townsend
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono ME, United States of America
- School of Biology and Ecology, University of Maine, Orono ME, United States of America
- * E-mail:
| |
Collapse
|
36
|
Zhu Q, Liu X, Glazier BJ, Krolick KN, Yang S, He J, Lo CC, Shi H. Differential Sympathetic Activation of Adipose Tissues by Brain-Derived Neurotrophic Factor. Biomolecules 2019; 9:biom9090452. [PMID: 31492038 PMCID: PMC6769916 DOI: 10.3390/biom9090452] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/17/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
Centrally administered brain-derived neurotrophic factor (BDNF) decreases body adiposity beyond what can be accounted for by decreased food intake, implying enhanced lipid metabolism by BDNF. Consistent with this notion, intracerebroventricular (icv) injection of BDNF in rats increased the expression of lipolytic enzymes in white adipose tissues (WAT) and increased circulating concentrations of lipolytic products without changing the levels of adrenal gland hormones. This suggests that central BDNF-induced lipid mobilization is likely due to sympathetic neural activation, rather than activation of the adrenocortical or adrenomedullary system. We hypothesized that BDNF activated sympathetic innervation of adipose tissues to regulate lipolysis. Rats with unilateral denervation of interscapular brown adipose tissue (BAT) and different WAT depots received icv injections of saline or BDNF. Both intact and denervated adipose tissues were exposed to the same circulating factors, but denervated adipose tissues did not receive neural signals. Norepinephrine (NE) turnover (NETO) of BAT and WAT was assessed as a measure of sympathetic activity. Findings revealed that central BDNF treatment induced a change in NETO in some but not all the adipose tissues tested. Specifically, greater NETO rates were found in BAT and gonadal epididymal WAT (EWAT), but not in inguinal WAT (IWAT) or retroperitoneal WAT (RWAT), of BDNF-treated rats compared to saline-treated rats. Furthermore, intact innervation was necessary for BDNF-induced NETO in BAT and EWAT. In addition, BDNF increased the expression of lipolytic enzymes in both intact and denervated EWAT and IWAT, suggesting that BDNF-induced WAT lipolysis was independent of intact innervation. To summarize, centrally administered BDNF selectively provoked sympathetic drives to BAT and EWAT that was dependent on intact innervation, while BDNF also increased lipolysis in a manner independent of intact innervation.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Xian Liu
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | | | | | - Shangyuwen Yang
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Jingyan He
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Chunmin C Lo
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Diabetes Institute, Ohio University, Athens, OH 45701, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
37
|
White JD, Dewal RS, Stanford KI. The beneficial effects of brown adipose tissue transplantation. Mol Aspects Med 2019; 68:74-81. [PMID: 31228478 DOI: 10.1016/j.mam.2019.06.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/20/2019] [Accepted: 06/18/2019] [Indexed: 01/02/2023]
Abstract
Obesity is a disease that results from an imbalance between energy intake and energy expenditure. Brown adipose tissue (BAT) is a potential therapeutic target to improve the comorbidities associated with obesity due to its inherent thermogenic capacity and its ability to improve glucose metabolism. Multiple studies have shown that activation of BAT using either pharmacological treatments or cold exposure had an acute effect to increase metabolic function and reduce adiposity. Recent preclinical investigations have explored whether increasing BAT mass or activation through transplantation models could improve glucose metabolism and metabolic health. Successful BAT transplantation models have shown improvements in glucose metabolism and insulin sensitivity, as well as reductions in body mass and decreased adiposity in recipients. BAT transplantation may confer its beneficial effects through several different mechanisms, including endocrine effects via the release of 'batokines'. More recent studies have demonstrated that beige and brown adipocytes isolated from human progenitor cells and transplanted into mouse models result in metabolic improvements similar to transplantation of whole BAT; this could represent a clinically translatable model. In this review we will discuss the impetus for both early and recent investigations utilizing BAT transplantation models, the outcomes of these studies, and review the mechanisms associated with the beneficial effects of BAT transplant to confer improvements in metabolic health.
Collapse
Affiliation(s)
- Joseph D White
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Revati S Dewal
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
38
|
Zhu Q, Glazier BJ, Hinkel BC, Cao J, Liu L, Liang C, Shi H. Neuroendocrine Regulation of Energy Metabolism Involving Different Types of Adipose Tissues. Int J Mol Sci 2019; 20:E2707. [PMID: 31159462 PMCID: PMC6600468 DOI: 10.3390/ijms20112707] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/17/2022] Open
Abstract
Despite tremendous research efforts to identify regulatory factors that control energy metabolism, the prevalence of obesity has been continuously rising, with nearly 40% of US adults being obese. Interactions between secretory factors from adipose tissues and the nervous system innervating adipose tissues play key roles in maintaining energy metabolism and promoting survival in response to metabolic challenges. It is currently accepted that there are three types of adipose tissues, white (WAT), brown (BAT), and beige (BeAT), all of which play essential roles in maintaining energy homeostasis. WAT mainly stores energy under positive energy balance, while it releases fuels under negative energy balance. Thermogenic BAT and BeAT dissipate energy as heat under cold exposure to maintain body temperature. Adipose tissues require neural and endocrine communication with the brain. A number of WAT adipokines and BAT batokines interact with the neural circuits extending from the brain to cooperatively regulate whole-body lipid metabolism and energy homeostasis. We review neuroanatomical, histological, genetic, and pharmacological studies in neuroendocrine regulation of adipose function, including lipid storage and mobilization of WAT, non-shivering thermogenesis of BAT, and browning of BeAT. Recent whole-tissue imaging and transcriptome analysis of differential gene expression in WAT and BAT yield promising findings to better understand the interaction between secretory factors and neural circuits, which represents a novel opportunity to tackle obesity.
Collapse
Affiliation(s)
- Qi Zhu
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Bradley J Glazier
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Benjamin C Hinkel
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Jingyi Cao
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Lin Liu
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Chun Liang
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Haifei Shi
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
39
|
Gomes A, Soares R, Costa R, Marino F, Cosentino M, Malagon MM, Ribeiro L. High-fat diet promotes adrenaline production by visceral adipocytes. Eur J Nutr 2019; 59:1105-1114. [PMID: 31011795 DOI: 10.1007/s00394-019-01971-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/15/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE Catecholamines (CA) play a major role in metabolism and immune response. Recent reports showing adipose tissue can synthetize CA enlighten new roles for these amines in obesity. This study aimed to evaluate the expression of both tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT) and CA content along preadipocytes differentiation, under normal and obesigenic conditions. METHODS 8-9 week-old male C57BL/6 mice were divided in two groups: one fed with a high-fat diet (HFD) and other with a standard diet (SD) for 20 weeks. Afterwards, both TH and PNMT expression, localization, and CA content in adipocytes, were evaluated. RESULTS qPCR results showed no changes for TH and PNMT expression during the differentiation process for visceral and subcutaneous preadipocytes from mice fed with SD. Comparing to SD, HFD increased TH gene expression of subcutaneous preadipocytes and PNMT gene expression of both visceral preadipocytes and adipocytes. HPLC-ED analyses revealed HFD increased visceral adipocytes noradrenaline intracellular content comparing with preadipocytes (p = 0.037). When compared with SD, HFD raised and decreased noradrenaline content, respectively, in visceral adipocytes (p = 0.004) and subcutaneous preadipocytes (p = 0.001). Along the differentiation process, HFD increased visceral adrenaline intracellular content comparing with SD (p < 0.001). HFD increased visceral comparing to subcutaneous adrenaline content for both preadipocytes (p = 0.004) and adipocytes (p = 0.001). CONCLUSIONS TH and PNMT expression in adipose tissue is differently modulated in visceral and subcutaneous adipose depots, and seems to depend on diet. Differences observed in visceral adipose CA handling in HFD-fed mice might uncover novel pharmacological/nutritional strategies against obesity and cardiovascular risk.
Collapse
Affiliation(s)
- Andreia Gomes
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Alameda Prof Hernâni Monteiro, 4200-319, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Raquel Soares
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Alameda Prof Hernâni Monteiro, 4200-319, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Raquel Costa
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Alameda Prof Hernâni Monteiro, 4200-319, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varèse, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varèse, Italy
| | - Maria M Malagon
- Department of Cell Biology, Physiology and Immunology, Instituto Maimonides de Investigación Biomédica (IMIBIC), Reina Sofia University Hospital, Av. Menéndez Pidal, 14004, Córdoba, Spain
| | - Laura Ribeiro
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Alameda Prof Hernâni Monteiro, 4200-319, Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
40
|
Herradon G, Ramos-Alvarez MP, Gramage E. Connecting Metainflammation and Neuroinflammation Through the PTN-MK-RPTPβ/ζ Axis: Relevance in Therapeutic Development. Front Pharmacol 2019; 10:377. [PMID: 31031625 PMCID: PMC6474308 DOI: 10.3389/fphar.2019.00377] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a common factor of pathologies such as obesity, type 2 diabetes or neurodegenerative diseases. Chronic inflammation is considered part of the pathogenic mechanisms of different disorders associated with aging. Interestingly, peripheral inflammation and the associated metabolic alterations not only facilitate insulin resistance and diabetes but also neurodegenerative disorders. Therefore, the identification of novel pathways, common to the development of these diseases, which modulate the immune response and signaling is key. It will provide highly relevant information to advance our knowledge of the multifactorial process of aging, and to establish new biomarkers and/or therapeutic targets to counteract the underlying chronic inflammatory processes. One novel pathway that regulates peripheral and central immune responses is triggered by the cytokines pleiotrophin (PTN) and midkine (MK), which bind its receptor, Receptor Protein Tyrosine Phosphatase (RPTP) β/ζ, and inactivate its phosphatase activity. In this review, we compile a growing body of knowledge suggesting that PTN and MK modulate the immune response and/or inflammation in different pathologies characterized by peripheral inflammation associated with insulin resistance, such as aging, and in central disorders characterized by overt neuroinflammation, such as neurodegenerative diseases and endotoxemia. Evidence strongly suggests that regulation of the PTN and MK signaling pathways may provide new therapeutic opportunities particularly in those neurological disorders characterized by increased PTN and/or MK cerebral levels and neuroinflammation. Importantly, we discuss existing therapeutics, and others being developed, that modulate these signaling pathways, and their potential use in pathologies characterized by overt neuroinflammation.
Collapse
Affiliation(s)
- Gonzalo Herradon
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - M Pilar Ramos-Alvarez
- Departmento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
41
|
Evans BA, Merlin J, Bengtsson T, Hutchinson DS. Adrenoceptors in white, brown, and brite adipocytes. Br J Pharmacol 2019; 176:2416-2432. [PMID: 30801689 DOI: 10.1111/bph.14631] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/28/2019] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Adrenoceptors play an important role in adipose tissue biology and physiology that includes regulating the synthesis and storage of triglycerides (lipogenesis), the breakdown of stored triglycerides (lipolysis), thermogenesis (heat production), glucose metabolism, and the secretion of adipocyte-derived hormones that can control whole-body energy homeostasis. These processes are regulated by the sympathetic nervous system through actions at different adrenoceptor subtypes expressed in adipose tissue depots. In this review, we have highlighted the role of adrenoceptor subtypes in white, brown, and brite adipocytes in both rodents and humans and have included detailed analysis of adrenoceptor expression in human adipose tissue and clonally derived adipocytes. We discuss important considerations when investigating adrenoceptor function in adipose tissue or adipocytes. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jon Merlin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
42
|
Chapelot D, Charlot K. Physiology of energy homeostasis: Models, actors, challenges and the glucoadipostatic loop. Metabolism 2019; 92:11-25. [PMID: 30500561 DOI: 10.1016/j.metabol.2018.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/25/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
Abstract
The aim of this review is to discuss the physiology of energy homeostasis (EH), which is a debated concept. Thus, we will see that the set-point theory is highly challenged and that other models integrating an anticipative component, such as energy allostasis, seem more relevant to experimental reports and life preservation. Moreover, the current obesity epidemic suggests that EH is poorly efficient in the modern human dietary environment. Non-homeostatic phenomena linked to hedonism and reward seem to profoundly impair EH. In this review, the apparent failed homeostatic responses to energy challenges such as exercise, cafeteria diet, overfeeding and diet-induced weight loss, as well as their putative determinants, are analyzed to highlight the mechanisms of EH. Then, the hormonal, neuronal, and metabolic factors of energy intake or energy expenditure are briefly presented. Last, this review focuses on the contributions of two of the most pivotal and often overlooked determinants of EH: the availability of endogenous energy and the pattern of energy intake. A glucoadipostatic loop model is finally proposed to link energy stored in adipose tissue to EH through changes in eating behavior via leptin and sympathetic nervous system activity.
Collapse
Affiliation(s)
- Didier Chapelot
- Université Paris 13, Centre de Recherche en Epidémiologie et Statistique, Equipe de Recherche en Epidémiologie Nutritionnelle (EREN), Inserm (U1153), Inra (U1125), Cnam, Bobigny, France.
| | - Keyne Charlot
- Institut de Recherche Biomédicale des Armées, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Département Environnements Opérationnels, Brétigny-sur-Orge, France
| |
Collapse
|
43
|
Blaszkiewicz M, Willows JW, Johnson CP, Townsend KL. The Importance of Peripheral Nerves in Adipose Tissue for the Regulation of Energy Balance. BIOLOGY 2019; 8:E10. [PMID: 30759876 PMCID: PMC6466238 DOI: 10.3390/biology8010010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/29/2022]
Abstract
Brown and white adipose tissues are essential for maintenance of proper energy balance and metabolic health. In order to function efficiently, these tissues require both endocrine and neural communication with the brain. Brown adipose tissue (BAT), as well as the inducible brown adipocytes that appear in white adipose tissue (WAT) after simulation, are thermogenic and energy expending. This uncoupling protein 1 (UCP1)-mediated process requires input from sympathetic nerves releasing norepinephrine. In addition to sympathetic noradrenergic signaling, adipose tissue contains sensory nerves that may be important for relaying fuel status to the brain. Chemical and surgical denervation studies of both WAT and BAT have clearly demonstrated the role of peripheral nerves in browning, thermogenesis, lipolysis, and adipogenesis. However, much is still unknown about which subtypes of nerves are present in BAT versus WAT, what nerve products are released from adipose nerves and how they act to mediate metabolic homeostasis, as well as which cell types in adipose are receiving synaptic input. Recent advances in whole-depot imaging and quantification of adipose nerve fibers, as well as other new research findings, have reinvigorated this field of research. This review summarizes the history of research into adipose innervation and brain⁻adipose communication, and also covers landmark and recent research on this topic to outline what we currently know and do not know about adipose tissue nerve supply and communication with the brain.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| | - Jake W Willows
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA.
| | - Cory P Johnson
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| | - Kristy L Townsend
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
44
|
Ali SB, Turner JJO, Fountain SJ. Constitutive P2Y 2 receptor activity regulates basal lipolysis in human adipocytes. J Cell Sci 2018; 131:jcs.221994. [PMID: 30333139 DOI: 10.1242/jcs.221994] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022] Open
Abstract
White adipocytes are key regulators of metabolic homeostasis, which release stored energy as free fatty acids via lipolysis. Adipocytes possess both basal and stimulated lipolytic capacity, but limited information exists regarding the molecular mechanisms that regulate basal lipolysis. Here, we describe a mechanism whereby autocrine purinergic signalling and constitutive P2Y2 receptor activation suppresses basal lipolysis in primary human in vitro-differentiated adipocytes. We found that human adipocytes possess cytoplasmic Ca2+ tone due to ATP secretion and constitutive P2Y2 receptor activation. Pharmacological antagonism or knockdown of P2Y2 receptors increases intracellular cAMP levels and enhances basal lipolysis. P2Y2 receptor antagonism works synergistically with phosphodiesterase inhibitors in elevating basal lipolysis, but is dependent upon adenylate cyclase activity. Mechanistically, we suggest that the increased Ca2+ tone exerts an anti-lipolytic effect by suppression of Ca2+-sensitive adenylate cyclase isoforms. We also observed that acute enhancement of basal lipolysis following P2Y2 receptor antagonism alters the profile of secreted adipokines leading to longer-term adaptive decreases in basal lipolysis. Our findings demonstrate that basal lipolysis and adipokine secretion are controlled by autocrine purinergic signalling in human adipocytes.
Collapse
Affiliation(s)
- Seema B Ali
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| | - Jeremy J O Turner
- Norfolk and Norwich University Hospital, Colney Lane, NR4 7TJ Norwich, UK
| | - Samuel J Fountain
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, NR4 7TJ Norwich, UK
| |
Collapse
|
45
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
46
|
Harris RBS. Denervation as a tool for testing sympathetic control of white adipose tissue. Physiol Behav 2018; 190:3-10. [PMID: 28694155 PMCID: PMC5758439 DOI: 10.1016/j.physbeh.2017.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 10/19/2022]
Abstract
This review summarizes the evidence derived from studies utilizing denervation procedures to demonstrate sympathetic control of white adipose tissue metabolism and body fat mass. A majority of the work demonstrating neural control of white fat was performed in the Bartness laboratory with Siberian hamsters as the predominant experimental model. These animals experience dramatic changes in body fat mass in response to changes in photoperiod, however, the mechanisms identified in hamsters have been reproduced or further elucidated by experiments with other animal models. Evidence for the role of sympathetic innervation contributing to the control of white adipocyte lipolysis and preadipocyte proliferation is summarized. In addition, evidence from denervation experiments for neural communication between different white fat depots as well as for a feedback control loop between sensory afferents from individual fat depots and sympathetic efferents to the same or distant white fat depots is discussed.
Collapse
Affiliation(s)
- Ruth B S Harris
- Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
47
|
Ailanen L, Vähätalo LH, Salomäki-Myftari H, Mäkelä S, Orpana W, Ruohonen ST, Savontaus E. Peripherally Administered Y 2-Receptor Antagonist BIIE0246 Prevents Diet-Induced Obesity in Mice With Excess Neuropeptide Y, but Enhances Obesity in Control Mice. Front Pharmacol 2018; 9:319. [PMID: 29674968 PMCID: PMC5895854 DOI: 10.3389/fphar.2018.00319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/20/2018] [Indexed: 12/27/2022] Open
Abstract
Neuropeptide Y (NPY) plays an important role in the regulation of energy homeostasis in the level of central and sympathetic nervous systems (SNSs). Genetic silencing of peripheral Y2-receptors have anti-obesity effects, but it is not known whether pharmacological blocking of peripheral Y2-receptors would similarly benefit energy homeostasis. The effects of a peripherally administered Y2-receptor antagonist were studied in healthy and energy-rich conditions with or without excess NPY. Genetically obese mice overexpressing NPY in brain noradrenergic nerves and SNS (OE-NPYDβH) represented the situation of elevated NPY levels, while wildtype (WT) mice represented the normal NPY levels. Specific Y2-receptor antagonist, BIIE0246, was administered (1.3 mg/kg/day, i.p.) for 2 or 4.5 weeks to OE-NPYDβH and WT mice feeding on chow or Western diet. Treatment with Y2-receptor antagonist increased body weight gain in both genotypes on chow diet and caused metabolic disturbances (e.g., hyperinsulinemia and hypercholesterolemia), especially in WT mice. During energy surplus (i.e., on Western diet), blocking of Y2-receptors induced obesity in WT mice, whereas OE-NPYDβH mice showed reduced fat mass gain, hepatic glycogen and serum cholesterol levels relative to body adiposity. Thus, it can be concluded that with normal NPY levels, peripheral Y2-receptor antagonist has no potential for treating obesity, but oppositely may even induce metabolic disorders. However, when energy-rich diet is combined with elevated NPY levels, e.g., stress combined with an unhealthy diet, Y2-receptor antagonism has beneficial effects on metabolic status.
Collapse
Affiliation(s)
- Liisa Ailanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Laura H Vähätalo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Henriikka Salomäki-Myftari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Satu Mäkelä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Wendy Orpana
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
48
|
Li QC, Li QF, Wang YL, Sun HL, Jiang ZY. Arcuate nucleus neurons are not essential for the preprandial peak in plasma ghrelin after neonatal monosodium glutamate treatment. Int J Mol Med 2018; 41:1635-1642. [PMID: 29328403 DOI: 10.3892/ijmm.2018.3365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/21/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to determine whether arcuate nucleus (ARC) lesions affect the ghrelin level in the plasma and the stomach in monosodium glutamate (MSG)‑treated mice. The aim of the present study was to investigate whether the ARC was destroyed in mice treated neonatally with MSG, and whether the ARC lesions affect the ghrelin level in the plasma and lipid mobilization in MSG‑treated mice. The results revealed that MSG led to a marked reduction in ARC cresyl violet staining, tyrosine hydroxylase-immunoreactive (IR) neurons and neuropeptide Y‑IR fibers, compared with saline controls. MSG‑treated mice exhibited significantly increased body mass compared with saline controls, and MSG treatment did not prevent food deprivation‑induced decrease in white adipose tissue mass compared with controls. Plasma ghrelin levels were significantly increased in MSG‑treated mice that were fasted for 48 h, compared with the levels prior to fasting and re‑feeding, and the preprandial peak of plasma ghrelin persisted in MSG‑treated mice. In summary, the ARC was not found to be essential for food deprivation‑induced lipid mobilization and preprandial peak in MSG‑treated mice. However, this finding does not mean that ARC neurons do not contribute to food sensing and lipid mobilization under normal conditions, as compensatory mechanisms may have emerged after the ablation of ARC neurons.
Collapse
Affiliation(s)
- Qing-Chun Li
- Department of Reproductive Medicine, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Qing-Feng Li
- Smart Division Gas, Qingdao iESLab Electronic Co., Ltd., Qingdao 266071, P.R. China
| | - Yan-Lin Wang
- Department of Reproductive Medicine, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Hong-Liang Sun
- Department of Reproductive Medicine, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, P.R. China
| | - Zheng-Yao Jiang
- Department of Physiology, Qingdao University Medical College, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
49
|
van der Spek R, Fliers E, la Fleur SE, Kalsbeek A. Daily Gene Expression Rhythms in Rat White Adipose Tissue Do Not Differ Between Subcutaneous and Intra-Abdominal Depots. Front Endocrinol (Lausanne) 2018; 9:206. [PMID: 29760682 PMCID: PMC5936761 DOI: 10.3389/fendo.2018.00206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/12/2018] [Indexed: 01/05/2023] Open
Abstract
White adipose tissue (WAT) is present in different depots throughout the body. Although all depots are exposed to systemic humoral signals, they are not functionally identical. Studies in clock gene knockout animals and in shift workers suggest that daily rhythmicity may play an important role in lipid metabolism. Differences in rhythmicity between fat depots might explain differences in depot function; therefore, we measured mRNA expression of clock genes and metabolic genes on a 3-h interval over a 24-h period in the subcutaneous inguinal depot and in the intra-abdominal perirenal, epididymal, and mesenteric depots of male Wistar rats. We analyzed rhythmicity using CircWave software. Additionally, we measured plasma concentrations of glucose, insulin, corticosterone, and leptin. The clock genes (Bmal1/Per2/Cry1/Cry2/RevErbα/DBP) showed robust daily gene expression rhythms, which did not vary between WAT depots. Metabolic gene expression rhythms (SREBP1c/PPARα/PPARγ/FAS/LPL/Glut4/HSL/CPT1b/leptin/visfatin/resistin) were more variable between depots. However, no distinct differences between intra-abdominal and subcutaneous rhythms were found. Concluding, specific fat depots are not associated with differences in clock gene expression rhythms and, therefore, do not provide a likely explanation for the differences in metabolic function between different fat depots.
Collapse
Affiliation(s)
- Rianne van der Spek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Rianne van der Spek,
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Susanne E. la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, Netherlands
| |
Collapse
|
50
|
Thomas MA, Tran V, Ryu V, Xue B, Bartness TJ. AgRP knockdown blocks long-term appetitive, but not consummatory, feeding behaviors in Siberian hamsters. Physiol Behav 2017; 190:61-70. [PMID: 29031552 DOI: 10.1016/j.physbeh.2017.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/24/2017] [Accepted: 10/06/2017] [Indexed: 12/25/2022]
Abstract
Arcuate hypothalamus-derived agouti-related protein (AgRP) and neuropeptide Y (NPY) are critical for maintaining energy homeostasis. Fasting markedly upregulates AgRP/NPY expression and circulating ghrelin, and exogenous ghrelin treatment robustly increases acute food foraging and food intake, and chronic food hoarding behaviors in Siberian hamsters. We previously demonstrated that 3rd ventricular AgRP injection robustly stimulates acute and chronic food hoarding, largely independent of food foraging and intake. By contrast, 3rd ventricular NPY injection increases food foraging, food intake, and food hoarding, but this effect is transient and gone by 24h post-injection. Because of this discrepancy in AgRP/NPY-induced ingestive behaviors, we tested whether selective knockdown of AgRP blocks fasting and ghrelin-induced increases in food hoarding. AgRP gene knockdown by a novel DICER small interfering RNA (AgRP-DsiRNA) blocked food-deprivation induced increases in AgRP expression, but had no effect on NPY expression. AgRP-DsiRNA attenuated acute (1day), and significantly decreased chronic (4-6days), food deprivation-induced increases in food hoarding. In addition, AgRP-DsiRNA treatment blocked exogenous ghrelin-induced increases in food hoarding through day 3, but had no effect on basal food foraging, food intake, or food hoarding prior to ghrelin treatment. Lastly, chronic AgRP knockdown had no effect on body mass, fat mass, or lean mass in either food deprived or ad libitum fed hamsters. These data collectively suggest that the prolonged increase in food hoarding behavior following energetic challenges, and food deprivation especially, is primarily regulated by downstream AgRP signaling.
Collapse
Affiliation(s)
- M Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Vy Tran
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Vitaly Ryu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA; Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA.
| | - Timothy J Bartness
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA; Neuroscience Institute, Georgia State University, Atlanta, GA, 30302, USA
| |
Collapse
|